Distribution, Identity and Putative Function of Fibroblast Growth Factor 10 Expressing Cells in the Developing and Adult Mouse Brain

Total Page:16

File Type:pdf, Size:1020Kb

Distribution, Identity and Putative Function of Fibroblast Growth Factor 10 Expressing Cells in the Developing and Adult Mouse Brain Distribution, identity and putative function of fibroblast growth factor 10 expressing cells in the developing and adult mouse brain A thesis submitted to the University of East Anglia for the degree of Doctor of Philosophy by Niels Haan April 2012 This copy of the thesis has been supplied on condition that anyone who consults it is understood to recognise that its copyright rest with the author and that no quotation from the thesis, not any information there from may be published without the author’s prior written consent. Abstract Neural stem cells in the adult mammalian brain have been extensively studied and well characterised. However, apart from the classical stem cell niches of the lateral ventricle and the hippocampus, other brain areas, including the hypothalamus and amygdala, are potential novel stem cell niches. These areas have previously been found to express fibroblast growth factor 10 (Fgf10), known to maintain stem cell niches in other tissues. Here the hypothalamus and amygdala are investigated as potential novel neural stem cell niches, and the possibility of the olfactory bulb as the target of a putative migratory stream from the amygdala is assessed. Using a combination of immunohistochemistry, BrdU incorporation assays, genetic lineage tracing and in vitro cultures, the properties of Fgf10 expressing cells were examined. The Fgf10 expressing cells of the hypothalamus, the tanycytes, were found to express stem cell markers, and are capable of division in vivo. This stem cell niche was found to be generated during development and continues to be present into adulthood. Although proliferation is highest in the early post-natal period, it continues into later life. The Fgf10+ lineage is able to generate neurons and glia both in vivo and in vitro. Given the location of newly generated Fgf10+ lineage derived neurons in the hypothalamus, a function in control of energy balance seems likely. In contrast, in the amygdala Fgf10+ cells do not appear to be stem cells, but mostly mature neurons. These do not express neural stem cell markers or incorporate BrdU. In the olfactory bulb, the majority of Fgf10+ cells, which are found in all layers, are mature neurons, of both a glutamatergic and GABAergic phenotype. Although the function of Fgf10 in these brain areas is as yet unknown, it marks a diverse population of cells, and is a candidate molecule to fulfil a number of different functions in these cells. 2 Acknowledgements First and foremost I must thank my supervisor, Mohammad K Hajihosseini for continued advice and support and for pushing me to achieve a great deal in my three years in Norwich. My secondary supervisors, Grant Wheeler and Uli Mayer, provided much useful input to my project. I am grateful to the Anatomical Society of Great Britain and Ireland for providing the funding for this project. In the lab, I must thank my Alaleh Najdi-Samiei for trustworthy and speedy genotyping and for making sure we always had ample supplies of anything we could think off, Christina Stratford and Irina Djacova for always being fun to be with and always willing to listen to me rant. Special thanks goes out to Timothy Goodman for being a good colleague, sounding board and for injecting what must at times have seemed millions of mice. Both in and out of the lab you have been great people to be with. I couldn’t have asked for better people to work with. Outside the UEA I sincerely thank Dr Saverio Bellusci for the Fgf10CreERT2 mouse line, and Dr Natalie Bronstein for the custom Fgf10 antibody. I would like to thank the Wheeler and Münsterberg lab members for never looking too bored during my lab meetings, and for always being fun company outside of the lab. The microscope support received from Paul Thomas and Alba Warn has always been useful. Special thanks goes out to the staff of the DMU, present and former, Simon Deakin, Rich Croft and Donna Hinds, for animal maintenance. In the BMRC I would like to single out the Mogensen lab for special thanks, for putting up with me continuously using their Western equipment and for much help, support and company. The rest of the people in the BMRC, former and present, have always been good friends and colleagues, and always put up with me asking to ‘borrow’ things or annoying them with small talk and gossip when trying to avoid doing work. I hope I have on occasion been useful in return as well. I must thank my parents and grandfather for financial support and apologize for seriously neglecting them whilst being all too busy in distant England. My greatest thanks go out to Jenny, Rob and Sally, for being the best friends, drinking buddies, and support group I could have wished for. I couldn’t have done it without you guys. 3 Index Abstract 2 Acknowledgments 3 Index 4 List of figures 8 List of tables 10 Chapter 1 – Introduction 11 1.1 – Adult neural stem cells 12 1.2 – Origin of adult neural stem cells 12 1.3 – Canonical niches for adult neural stem cells 14 1.3.1 Subventricular zone of the lateral ventricle 14 1.3.2 Subgranular zone of the dentate gyrus 17 1.4 – Potential new niches for adult neural stem cells 18 1.5 – Selfrenewal of adult neural stem cells 19 1.6 – Neuroblast migration 21 1.7 – Differentiation of neural stem cells 24 1.8 – Fibroblast growth factors 26 1.9 – Fgf receptors 26 1.10 – Fgf expression and function in the developing and adult nervous 30 system 1.10.1 Fgf1 subfamily (Fgf1 and 2) 30 1.10.2 Fgf4 subfamily (Fgf4, 5 and 6) 31 1.10.3 Fgf7 subfamily (Fgf3, 7, 10 and 22) 31 1.10.4 Fgf8 subfamily (Fgf8, 17 and 18) 32 1.10.5 Fgf9 subfamily (Fgf9, 16 and 20) 32 1.10.6 iFgf subfamily (Fgfs 11-14) 33 1.10.7 hFgf subfamily (Fgfs 15/19, 21 and 23 33 1.11 – Fgf10 34 1.12 – Fgf10 knockouts 34 1.13 – Mutations in Fgf10 and disease in humans 35 1.14 – Roles of Fgf10 in tissue development. 35 1.14.1 Lung 36 1.14.2 Stomach 37 1.14.3 Pancreas 37 1.14.4 Limbs 38 1.14.5 Adipose tissue 38 1.14.6 Teeth 39 1.14.7 Small intestine 39 1.14.8 Liver 39 1.15 – Expression and functions Fgf10 in the brain 40 1.16 – Aims 43 4 Chapter 2 – Materials and methods 45 2.1 – Mouse lines 46 2.1.1 Fgf10lacZ 46 2.1.2 NestincreERT2 46 2.1.3 Fgf10creERT2 46 2.1.4 ROSA26YFP/ROSA26RFP/ROSA26LacZ 48 2.1.5 Fgf10 KO 48 2.1.6 Fgf10CreERT2::ROSA26Tomato 48 2.6.7 Breeding and genotyping 48 2.2 - Animal treatments 50 2.2.1 Time mating 50 2.2.2 BrdU administration 50 2.2.3 Tamoxifen administration 50 2.3 - Tissue processing 50 2.4 - Sectioning 51 2.5 - Immunohistochemistry 52 2.5.1 Antigen retrievals 52 2.5.2 Cryostat section staining 53 2.5.3 Vibratome section staining 54 2.6 - Cell culture 56 2.6.1 Culture surface coatings 56 2.6.2 Neurosphere culture 57 2.6.3 Primary neurons 59 2.6.4 Primary astrocytes 59 2.6.5 Cell lines 60 2.6.6 Xgal staining on cells 60 2.7 - Immunocytochemistry 60 2.8 - Microscopy 61 2.9 - Flow cytometry 61 2.10 – Reverse transcriptase PCR 62 2.10.1 Sample preparation 62 2.10.2 RT-PCR 63 2.11 - Western blot 64 2.11.1 Sample preparation 64 2.11.2 SDS-PAGE and transfer 64 2.11.3 Detection 65 2.11.4 ECL detection 66 2.11.5 Quantification 66 2.12 Statistical analysis 66 Chapter 3 – Optimisation of detection of FGF10 protein in 67 immunohistochemistry and Western blot 3.1 – Introduction 68 3.2 – Results 70 3.2.1 Commercial anti-FGF10 antibodies do not work in 70 immunohistochemistry 3.2.2 Custom anti-FGF10 does not appear to be specific for FGF10 70 3.2.3 Custom anti-FGF10 does detect Fgf10 in western blot, but is 71 not specific 5 3.2.4 Specificity can be somewhat improved using 74 unconventional blotting techniques 3.2.5 Custom antibody may be used for quantification of FGF10 77 protein levels 3.3 Discussion 78 Chapter 4 - Distribution and phenotype of Fgf10-lacZ expressing cells in the 80 developing and adult hypothalamus 4.1 – Introduction 81 4.2 – Results 85 4.2.1 Fgf10-lacZ positive population is dynamic over time. 85 4.2.2 Fgf10-lacZ positive cells are located across the adult 85 hypothalamus 4.2.3 Fgf10-lacZ expressing cells are located mostly in the arcuate 88 nucleus 4.2.4 A heterogeneous population of tanycytes expresses neural 88 stem cell markers 4.2.5 Fgf10-lacZ positive cells express FgfR1-IIIc and 3-IIIc, but not 91 FgfR2 or FgfR4 4.2.6 Expression pattern of Fgf10-lacZ is largely set up before 93 birth 4.2.7 The Fgf10 expressing lineage contributes to embryonic 96 hypothalamic neurogenesis. 4.2.8 Fgf10-lacZ+ cells contribute neurons and glia to the 96 hypothalamic parenchyma 4.3 - Discussion 100 Chapter 5 - In vivo analysis of cell proliferation and genetic lineage tracing in 104 the hypothalamus 5.1 – Introduction 105 5.2 - Results 109 5.2.1 Ki67 labeling shows limited proliferation in the adult 109 hypothalamus 5.2.2 Cumulative BrdU administration shows widespread 109 proliferation, including a subset of Fgf10-lacZ+ cells 5.2.3 Both total and Fgf10-lacZ expressing BrdU incorporating 112 populations decrease with age 5.2.4 Majority of Fgf10-lacZ+ cells do not divide after cessation of 112 embryonic neurogenesis 5.2.5 Inducible nestinCreERT2 animals are not suitable for adult 115 lineage tracing 5.2.6 Inducible genetic lineage tracing confirms Fgf10 expression 115 in the hypothalamus 5.2.7 Recombined cells do not express glial markers 119 5.2.8 Direct lineage tracing shows Fgf10 expressing cells generate 119 hypothalamic neurons 5.2.9 Embryonic induction of Fgf10CreERT2::ROSA26lacZ animals 119 shows Fgf10 distribution 6 5.1.10 Fgf10CreERT2::Rosa26Tomato mice confirms lineage tracing 123 results 5.1.11 Fgf10CreERT2::Rosa26Tomato mice show more widespread 123 recombination 5.3
Recommended publications
  • DNA Methylation of GHSR, GNG4, HOXD9 and SALL3 Is a Common Epigenetic Alteration in Thymic Carcinoma
    INTERNATIONAL JOURNAL OF ONCOLOGY 56: 315-326, 2020 DNA methylation of GHSR, GNG4, HOXD9 and SALL3 is a common epigenetic alteration in thymic carcinoma REINA KISHIBUCHI1, KAZUYA KONDO1, SHIHO SOEJIMA1, MITSUHIRO TSUBOI2, KOICHIRO KAJIURA2, YUKIKIYO KAWAKAMI2, NAOYA KAWAKITA2, TORU SAWADA2, HIROAKI TOBA2, MITSUTERU YOSHIDA2, HIROMITSU TAKIZAWA2 and AKIRA TANGOKU2 1Department of Oncological Medical Services, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8509; 2Department of Thoracic, Endocrine Surgery and Oncology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8503, Japan Received August 17, 2019; Accepted October 25, 2019 DOI: 10.3892/ijo.2019.4915 Abstract. Thymic epithelial tumors comprise thymoma, promoter methylation of the 4 genes was not significantly thymic carcinoma and neuroendocrine tumors of the thymus. higher in advanced-stage tumors than in early-stage tumors in Recent studies have revealed that the incidence of somatic all thymic epithelial tumors. Among the 4 genes, relapse-free non‑synonymous mutations is significantly higher in thymic survival was significantly worse in tumors with a higher DNA carcinoma than in thymoma. However, limited information methylation than in those with a lower DNA methylation in all is currently available on epigenetic alterations in these types thymic epithelial tumors. Moreover, relapse-free survival was of cancer. In this study, we thus performed genome-wide significantly worse in thymomas with a higher DNA methyla- screening of aberrantly methylated CpG islands in thymoma tion of HOXD9 and SALL3 than in those with a lower DNA and thymic carcinoma using Illumina HumanMethylation450 methylation. On the whole, the findings of this study indicated K BeadChip. We identified 92 CpG islands significantly that the promoter methylation of cancer-related genes was hypermethylated in thymic carcinoma in relation to thymoma significantly higher in thymic carcinoma than in thymoma and and selected G protein subunit gamma 4 (GNG4), growth the thymus.
    [Show full text]
  • Spatially Heterogeneous Choroid Plexus Transcriptomes Encode Positional Identity and Contribute to Regional CSF Production
    The Journal of Neuroscience, March 25, 2015 • 35(12):4903–4916 • 4903 Development/Plasticity/Repair Spatially Heterogeneous Choroid Plexus Transcriptomes Encode Positional Identity and Contribute to Regional CSF Production Melody P. Lun,1,3 XMatthew B. Johnson,2 Kevin G. Broadbelt,1 Momoko Watanabe,4 Young-jin Kang,4 Kevin F. Chau,1 Mark W. Springel,1 Alexandra Malesz,1 Andre´ M.M. Sousa,5 XMihovil Pletikos,5 XTais Adelita,1,6 Monica L. Calicchio,1 Yong Zhang,7 Michael J. Holtzman,7 Hart G.W. Lidov,1 XNenad Sestan,5 Hanno Steen,1 XEdwin S. Monuki,4 and Maria K. Lehtinen1 1Department of Pathology, and 2Division of Genetics, Boston Children’s Hospital, Boston, Massachusetts 02115, 3Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts 02118, 4Department of Pathology and Laboratory Medicine, University of California Irvine School of Medicine, Irvine, California 92697, 5Department of Neurobiology and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, Connecticut 06510, 6Department of Biochemistry, Federal University of Sa˜o Paulo, Sa˜o Paulo 04039, Brazil, and 7Pulmonary and Critical Care Medicine, Department of Medicine, Washington University, St Louis, Missouri 63110 A sheet of choroid plexus epithelial cells extends into each cerebral ventricle and secretes signaling factors into the CSF. To evaluate whether differences in the CSF proteome across ventricles arise, in part, from regional differences in choroid plexus gene expression, we defined the transcriptome of lateral ventricle (telencephalic) versus fourth ventricle (hindbrain) choroid plexus. We find that positional identitiesofmouse,macaque,andhumanchoroidplexiderivefromgeneexpressiondomainsthatparalleltheiraxialtissuesoforigin.We thenshowthatmolecularheterogeneitybetweentelencephalicandhindbrainchoroidplexicontributestoregion-specific,age-dependent protein secretion in vitro.
    [Show full text]
  • Table S1 the Four Gene Sets Derived from Gene Expression Profiles of Escs and Differentiated Cells
    Table S1 The four gene sets derived from gene expression profiles of ESCs and differentiated cells Uniform High Uniform Low ES Up ES Down EntrezID GeneSymbol EntrezID GeneSymbol EntrezID GeneSymbol EntrezID GeneSymbol 269261 Rpl12 11354 Abpa 68239 Krt42 15132 Hbb-bh1 67891 Rpl4 11537 Cfd 26380 Esrrb 15126 Hba-x 55949 Eef1b2 11698 Ambn 73703 Dppa2 15111 Hand2 18148 Npm1 11730 Ang3 67374 Jam2 65255 Asb4 67427 Rps20 11731 Ang2 22702 Zfp42 17292 Mesp1 15481 Hspa8 11807 Apoa2 58865 Tdh 19737 Rgs5 100041686 LOC100041686 11814 Apoc3 26388 Ifi202b 225518 Prdm6 11983 Atpif1 11945 Atp4b 11614 Nr0b1 20378 Frzb 19241 Tmsb4x 12007 Azgp1 76815 Calcoco2 12767 Cxcr4 20116 Rps8 12044 Bcl2a1a 219132 D14Ertd668e 103889 Hoxb2 20103 Rps5 12047 Bcl2a1d 381411 Gm1967 17701 Msx1 14694 Gnb2l1 12049 Bcl2l10 20899 Stra8 23796 Aplnr 19941 Rpl26 12096 Bglap1 78625 1700061G19Rik 12627 Cfc1 12070 Ngfrap1 12097 Bglap2 21816 Tgm1 12622 Cer1 19989 Rpl7 12267 C3ar1 67405 Nts 21385 Tbx2 19896 Rpl10a 12279 C9 435337 EG435337 56720 Tdo2 20044 Rps14 12391 Cav3 545913 Zscan4d 16869 Lhx1 19175 Psmb6 12409 Cbr2 244448 Triml1 22253 Unc5c 22627 Ywhae 12477 Ctla4 69134 2200001I15Rik 14174 Fgf3 19951 Rpl32 12523 Cd84 66065 Hsd17b14 16542 Kdr 66152 1110020P15Rik 12524 Cd86 81879 Tcfcp2l1 15122 Hba-a1 66489 Rpl35 12640 Cga 17907 Mylpf 15414 Hoxb6 15519 Hsp90aa1 12642 Ch25h 26424 Nr5a2 210530 Leprel1 66483 Rpl36al 12655 Chi3l3 83560 Tex14 12338 Capn6 27370 Rps26 12796 Camp 17450 Morc1 20671 Sox17 66576 Uqcrh 12869 Cox8b 79455 Pdcl2 20613 Snai1 22154 Tubb5 12959 Cryba4 231821 Centa1 17897
    [Show full text]
  • Watsonjn2018.Pdf (1.780Mb)
    UNIVERSITY OF CENTRAL OKLAHOMA Edmond, Oklahoma Department of Biology Investigating Differential Gene Expression in vivo of Cardiac Birth Defects in an Avian Model of Maternal Phenylketonuria A THESIS SUBMITTED TO THE GRADUATE FACULTY In partial fulfillment of the requirements For the degree of MASTER OF SCIENCE IN BIOLOGY By Jamie N. Watson Edmond, OK June 5, 2018 J. Watson/Dr. Nikki Seagraves ii J. Watson/Dr. Nikki Seagraves Acknowledgements It is difficult to articulate the amount of gratitude I have for the support and encouragement I have received throughout my master’s thesis. Many people have added value and support to my life during this time. I am thankful for the education, experience, and friendships I have gained at the University of Central Oklahoma. First, I would like to thank Dr. Nikki Seagraves for her mentorship and friendship. I lucked out when I met her. I have enjoyed working on this project and I am very thankful for her support. I would like thank Thomas Crane for his support and patience throughout my master’s degree. I would like to thank Dr. Shannon Conley for her continued mentorship and support. I would like to thank Liz Bullen and Dr. Eric Howard for their training and help on this project. I would like to thank Kristy Meyer for her friendship and help throughout graduate school. I would like to thank my committee members Dr. Robert Brennan and Dr. Lilian Chooback for their advisement on this project. Also, I would like to thank the biology faculty and staff. I would like to thank the Seagraves lab members: Jailene Canales, Kayley Pate, Mckayla Muse, Grace Thetford, Kody Harvey, Jordan Guffey, and Kayle Patatanian for their hard work and support.
    [Show full text]
  • 4-6 Weeks Old Female C57BL/6 Mice Obtained from Jackson Labs Were Used for Cell Isolation
    Methods Mice: 4-6 weeks old female C57BL/6 mice obtained from Jackson labs were used for cell isolation. Female Foxp3-IRES-GFP reporter mice (1), backcrossed to B6/C57 background for 10 generations, were used for the isolation of naïve CD4 and naïve CD8 cells for the RNAseq experiments. The mice were housed in pathogen-free animal facility in the La Jolla Institute for Allergy and Immunology and were used according to protocols approved by the Institutional Animal Care and use Committee. Preparation of cells: Subsets of thymocytes were isolated by cell sorting as previously described (2), after cell surface staining using CD4 (GK1.5), CD8 (53-6.7), CD3ε (145- 2C11), CD24 (M1/69) (all from Biolegend). DP cells: CD4+CD8 int/hi; CD4 SP cells: CD4CD3 hi, CD24 int/lo; CD8 SP cells: CD8 int/hi CD4 CD3 hi, CD24 int/lo (Fig S2). Peripheral subsets were isolated after pooling spleen and lymph nodes. T cells were enriched by negative isolation using Dynabeads (Dynabeads untouched mouse T cells, 11413D, Invitrogen). After surface staining for CD4 (GK1.5), CD8 (53-6.7), CD62L (MEL-14), CD25 (PC61) and CD44 (IM7), naïve CD4+CD62L hiCD25-CD44lo and naïve CD8+CD62L hiCD25-CD44lo were obtained by sorting (BD FACS Aria). Additionally, for the RNAseq experiments, CD4 and CD8 naïve cells were isolated by sorting T cells from the Foxp3- IRES-GFP mice: CD4+CD62LhiCD25–CD44lo GFP(FOXP3)– and CD8+CD62LhiCD25– CD44lo GFP(FOXP3)– (antibodies were from Biolegend). In some cases, naïve CD4 cells were cultured in vitro under Th1 or Th2 polarizing conditions (3, 4).
    [Show full text]
  • Genome-Wide DNA Methylation Analysis of KRAS Mutant Cell Lines Ben Yi Tew1,5, Joel K
    www.nature.com/scientificreports OPEN Genome-wide DNA methylation analysis of KRAS mutant cell lines Ben Yi Tew1,5, Joel K. Durand2,5, Kirsten L. Bryant2, Tikvah K. Hayes2, Sen Peng3, Nhan L. Tran4, Gerald C. Gooden1, David N. Buckley1, Channing J. Der2, Albert S. Baldwin2 ✉ & Bodour Salhia1 ✉ Oncogenic RAS mutations are associated with DNA methylation changes that alter gene expression to drive cancer. Recent studies suggest that DNA methylation changes may be stochastic in nature, while other groups propose distinct signaling pathways responsible for aberrant methylation. Better understanding of DNA methylation events associated with oncogenic KRAS expression could enhance therapeutic approaches. Here we analyzed the basal CpG methylation of 11 KRAS-mutant and dependent pancreatic cancer cell lines and observed strikingly similar methylation patterns. KRAS knockdown resulted in unique methylation changes with limited overlap between each cell line. In KRAS-mutant Pa16C pancreatic cancer cells, while KRAS knockdown resulted in over 8,000 diferentially methylated (DM) CpGs, treatment with the ERK1/2-selective inhibitor SCH772984 showed less than 40 DM CpGs, suggesting that ERK is not a broadly active driver of KRAS-associated DNA methylation. KRAS G12V overexpression in an isogenic lung model reveals >50,600 DM CpGs compared to non-transformed controls. In lung and pancreatic cells, gene ontology analyses of DM promoters show an enrichment for genes involved in diferentiation and development. Taken all together, KRAS-mediated DNA methylation are stochastic and independent of canonical downstream efector signaling. These epigenetically altered genes associated with KRAS expression could represent potential therapeutic targets in KRAS-driven cancer. Activating KRAS mutations can be found in nearly 25 percent of all cancers1.
    [Show full text]
  • Emx2 Is Required for Growth of the Hippocampus but Not for Hippocampal Field Specification
    The Journal of Neuroscience, April 1, 2000, 20(7):2618–2625 Emx2 Is Required for Growth of the Hippocampus But Not for Hippocampal Field Specification Shubha Tole,1 Guy Goudreau,2 Stavroula Assimacopoulos,1 and Elizabeth A. Grove1 1Department of Neurobiology, Pharmacology, and Physiology, University of Chicago, Chicago, Illinois 60637, and 2Max Planck Institute of Biophysical Chemistry, D-37077 Goettingen, Germany The vertebrate Emx genes are expressed in a nested pattern in positioned in the Emx2 mutant. In particular, a dentate cell early embryonic cerebral cortex, such that a medial strip of population is generated, although it fails to form a morpholog- cortex expresses Emx2 but not Emx1. This pattern suggests ical gyrus. This failure may be part of a more widespread medial that Emx genes could play a role in specifying different areas or cortical defect in the mutant. Examination of cortical cell pro- fields of the cortex along the mediolateral axis. Such a role has liferation and differentiation indicates a disruption of the matu- been supported by the observation that in mice lacking func- ration of the medial cortex in the absence of Emx2. Thus, Emx2 tional Emx2 the hippocampus is shrunken and the most medial is required for normal growth and maturation of the hippocam- field of the cortex, the hippocampal dentate gyrus, appears by pus but not for the specification of cells to particular hippocam- cytoarchitecture to be missing (Pellegrini et al., 1996; Yoshida et pal field identities. al., 1997). Use of region-specific molecular markers shows, Key words: Emx2; hippocampus; patterning; specification; however, that hippocampal fields are specified and correctly cortical maturation; cortical hem The hippocampus, like the rest of the cerebral cortex, is divided Reports that specific mutations lead to morphological defects in into cytoarchitectonic areas or fields (Nauta and Feirtag, 1986).
    [Show full text]
  • Single Cell Regulatory Landscape of the Mouse Kidney Highlights Cellular Differentiation Programs and Disease Targets
    ARTICLE https://doi.org/10.1038/s41467-021-22266-1 OPEN Single cell regulatory landscape of the mouse kidney highlights cellular differentiation programs and disease targets Zhen Miao 1,2,3,8, Michael S. Balzer 1,2,8, Ziyuan Ma 1,2,8, Hongbo Liu1,2, Junnan Wu 1,2, Rojesh Shrestha 1,2, Tamas Aranyi1,2, Amy Kwan4, Ayano Kondo 4, Marco Pontoglio 5, Junhyong Kim6, ✉ Mingyao Li 7, Klaus H. Kaestner2,4 & Katalin Susztak 1,2,4 1234567890():,; Determining the epigenetic program that generates unique cell types in the kidney is critical for understanding cell-type heterogeneity during tissue homeostasis and injury response. Here, we profile open chromatin and gene expression in developing and adult mouse kidneys at single cell resolution. We show critical reliance of gene expression on distal regulatory elements (enhancers). We reveal key cell type-specific transcription factors and major gene- regulatory circuits for kidney cells. Dynamic chromatin and expression changes during nephron progenitor differentiation demonstrates that podocyte commitment occurs early and is associated with sustained Foxl1 expression. Renal tubule cells follow a more complex differentiation, where Hfn4a is associated with proximal and Tfap2b with distal fate. Mapping single nucleotide variants associated with human kidney disease implicates critical cell types, developmental stages, genes, and regulatory mechanisms. The single cell multi-omics atlas reveals key chromatin remodeling events and gene expression dynamics associated with kidney development. 1 Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA. 2 Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
    [Show full text]
  • Epigenetic Services Citations
    Active Motif Epigenetic Services Publications The papers below contain data generated by Active Motif’s Epigenetic Services team. To learn more about our services, please give us a call or visit us at www.activemotif.com/services. Technique Target Journal Year Reference Justin C. Boucher et al. CD28 Costimulatory Domain- ATAC-Seq, Cancer Immunol. Targeted Mutations Enhance Chimeric Antigen Receptor — 2021 RNA-Seq Res. T-cell Function. Cancer Immunol. Res. doi: 10.1158/2326- 6066.CIR-20-0253. Satvik Mareedu et al. Sarcolipin haploinsufficiency Am. J. Physiol. prevents dystrophic cardiomyopathy in mdx mice. RNA-Seq — Heart Circ. 2021 Am J Physiol Heart Circ Physiol. doi: 10.1152/ Physiol. ajpheart.00601.2020. Gabi Schutzius et al. BET bromodomain inhibitors regulate Nature Chemical ChIP-Seq BRD4 2021 keratinocyte plasticity. Nat. Chem. Biol. doi: 10.1038/ Biology s41589-020-00716-z. Siyun Wang et al. cMET promotes metastasis and ChIP-qPCR FOXO3 J. Cell Physiol. 2021 epithelial-mesenchymal transition in colorectal carcinoma by repressing RKIP. J. Cell Physiol. doi: 10.1002/jcp.30142. Sonia Iyer et al. Genetically Defined Syngeneic Mouse Models of Ovarian Cancer as Tools for the Discovery of ATAC-Seq — Cancer Discovery 2021 Combination Immunotherapy. Cancer Discov. doi: doi: 10.1158/2159-8290 Vinod Krishna et al. Integration of the Transcriptome and Genome-Wide Landscape of BRD2 and BRD4 Binding BRD2, BRD4, RNA Motifs Identifies Key Superenhancer Genes and Reveals ChIP-Seq J. Immunol. 2021 Pol II the Mechanism of Bet Inhibitor Action in Rheumatoid Arthritis Synovial Fibroblasts. J. Immunol. doi: doi: 10.4049/ jimmunol.2000286. Daniel Haag et al.
    [Show full text]
  • The Role of Emx1 and Emx2 in the Developing Chick Telencephalon
    The Role of Emx1 and Emx2 in the developing chick telencephalon Dissertation Zur Erlangung des Doktorgrades der Naturwissenschaften (Dr.rer.nat.) der Fakultät für Biologie der Ludwig-Maximilian-Universität München Angefertigt am Max-Planck-Institut für Neurobiologie in der Arbeitsgruppe Neuronale Spezifizierung und in der GSF am Institut für Stammzellforschung Julia von Frowein München, Dezember 2004 1. Gutachter: Prof.Dr. Magdalena Götz 2. Gutachter: Prof.Dr. George Boyan eingereicht am 20.12.2004 Tag der mündlichen Prüfung: 26.4.2005 If the brain were so simple we could understand it, we would be so simple we couldn't. Lyall Watson Table of content 1 Table of content 1 Table of content...........................................................................................1 2 Abstract ........................................................................................................5 3 Zusammenfassung......................................................................................6 4 Introduction..................................................................................................8 4.1 General development of the regions of the central nervous system ....................................8 4.2 Patterning and regionalization .............................................................................................8 4.3 Regions of the forebrain.....................................................................................................10 4.4 Migration............................................................................................................................12
    [Show full text]
  • Supplementary Table 1
    Supplementary Table 1. 492 genes are unique to 0 h post-heat timepoint. The name, p-value, fold change, location and family of each gene are indicated. Genes were filtered for an absolute value log2 ration 1.5 and a significance value of p ≤ 0.05. Symbol p-value Log Gene Name Location Family Ratio ABCA13 1.87E-02 3.292 ATP-binding cassette, sub-family unknown transporter A (ABC1), member 13 ABCB1 1.93E-02 −1.819 ATP-binding cassette, sub-family Plasma transporter B (MDR/TAP), member 1 Membrane ABCC3 2.83E-02 2.016 ATP-binding cassette, sub-family Plasma transporter C (CFTR/MRP), member 3 Membrane ABHD6 7.79E-03 −2.717 abhydrolase domain containing 6 Cytoplasm enzyme ACAT1 4.10E-02 3.009 acetyl-CoA acetyltransferase 1 Cytoplasm enzyme ACBD4 2.66E-03 1.722 acyl-CoA binding domain unknown other containing 4 ACSL5 1.86E-02 −2.876 acyl-CoA synthetase long-chain Cytoplasm enzyme family member 5 ADAM23 3.33E-02 −3.008 ADAM metallopeptidase domain Plasma peptidase 23 Membrane ADAM29 5.58E-03 3.463 ADAM metallopeptidase domain Plasma peptidase 29 Membrane ADAMTS17 2.67E-04 3.051 ADAM metallopeptidase with Extracellular other thrombospondin type 1 motif, 17 Space ADCYAP1R1 1.20E-02 1.848 adenylate cyclase activating Plasma G-protein polypeptide 1 (pituitary) receptor Membrane coupled type I receptor ADH6 (includes 4.02E-02 −1.845 alcohol dehydrogenase 6 (class Cytoplasm enzyme EG:130) V) AHSA2 1.54E-04 −1.6 AHA1, activator of heat shock unknown other 90kDa protein ATPase homolog 2 (yeast) AK5 3.32E-02 1.658 adenylate kinase 5 Cytoplasm kinase AK7
    [Show full text]
  • Strand Breaks for P53 Exon 6 and 8 Among Different Time Course of Folate Depletion Or Repletion in the Rectosigmoid Mucosa
    SUPPLEMENTAL FIGURE COLON p53 EXONIC STRAND BREAKS DURING FOLATE DEPLETION-REPLETION INTERVENTION Supplemental Figure Legend Strand breaks for p53 exon 6 and 8 among different time course of folate depletion or repletion in the rectosigmoid mucosa. The input of DNA was controlled by GAPDH. The data is shown as ΔCt after normalized to GAPDH. The higher ΔCt the more strand breaks. The P value is shown in the figure. SUPPLEMENT S1 Genes that were significantly UPREGULATED after folate intervention (by unadjusted paired t-test), list is sorted by P value Gene Symbol Nucleotide P VALUE Description OLFM4 NM_006418 0.0000 Homo sapiens differentially expressed in hematopoietic lineages (GW112) mRNA. FMR1NB NM_152578 0.0000 Homo sapiens hypothetical protein FLJ25736 (FLJ25736) mRNA. IFI6 NM_002038 0.0001 Homo sapiens interferon alpha-inducible protein (clone IFI-6-16) (G1P3) transcript variant 1 mRNA. Homo sapiens UDP-N-acetyl-alpha-D-galactosamine:polypeptide N-acetylgalactosaminyltransferase 15 GALNTL5 NM_145292 0.0001 (GALNT15) mRNA. STIM2 NM_020860 0.0001 Homo sapiens stromal interaction molecule 2 (STIM2) mRNA. ZNF645 NM_152577 0.0002 Homo sapiens hypothetical protein FLJ25735 (FLJ25735) mRNA. ATP12A NM_001676 0.0002 Homo sapiens ATPase H+/K+ transporting nongastric alpha polypeptide (ATP12A) mRNA. U1SNRNPBP NM_007020 0.0003 Homo sapiens U1-snRNP binding protein homolog (U1SNRNPBP) transcript variant 1 mRNA. RNF125 NM_017831 0.0004 Homo sapiens ring finger protein 125 (RNF125) mRNA. FMNL1 NM_005892 0.0004 Homo sapiens formin-like (FMNL) mRNA. ISG15 NM_005101 0.0005 Homo sapiens interferon alpha-inducible protein (clone IFI-15K) (G1P2) mRNA. SLC6A14 NM_007231 0.0005 Homo sapiens solute carrier family 6 (neurotransmitter transporter) member 14 (SLC6A14) mRNA.
    [Show full text]